regulation of apoptosis-associated lysosomal membrane ...315254/fulltext01.pdf · micrographs (c),...

33
Linköping University Post Print Regulation of apoptosis-associated lysosomal membrane permeabilization Ann-Charlotte Johansson, Hanna Appelqvist, Cathrine Nilsson, Katarina Kågedal, Karin Roberg and Karin Öllinger N.B.: When citing this work, cite the original article. The original publication is available at www.springerlink.com: Ann-Charlotte Johansson, Hanna Appelqvist, Cathrine Nilsson, Katarina Kågedal, Karin Roberg and Karin Öllinger, Regulation of apoptosis-associated lysosomal membrane permeabilization, 2010, APOPTOSIS, (15), 5, 527-540. http://dx.doi.org/10.1007/s10495-009-0452-5 Copyright: Springer Science Business Media http://www.springerlink.com/ Postprint available at: Linköping University Electronic Press http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-55061

Upload: others

Post on 27-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

Linköping University Post Print

Regulation of apoptosis-associated lysosomal

membrane permeabilization

Ann-Charlotte Johansson, Hanna Appelqvist, Cathrine Nilsson, Katarina Kågedal,

Karin Roberg and Karin Öllinger

N.B.: When citing this work, cite the original article.

The original publication is available at www.springerlink.com:

Ann-Charlotte Johansson, Hanna Appelqvist, Cathrine Nilsson, Katarina Kågedal, Karin

Roberg and Karin Öllinger, Regulation of apoptosis-associated lysosomal membrane

permeabilization, 2010, APOPTOSIS, (15), 5, 527-540.

http://dx.doi.org/10.1007/s10495-009-0452-5

Copyright: Springer Science Business Media

http://www.springerlink.com/

Postprint available at: Linköping University Electronic Press

http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-55061

Page 2: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

REGULATION OF APOPTOSIS-ASSOCIATED LYSOSOMAL

MEMBRANE PERMEABILIZATION

Ann-CharlotteJohansson1, Hanna Appelqvist2, Cathrine Nilsson1,2,

Katarina Kågedal2, Karin Roberg1,3, Karin Öllinger2

1 Division of Otorhinolaryngology, Linköping University Hospital, Linköping, Sweden

2 Division of Experimental Pathology, Department of Clinical and Experimental Medicine,

Linköping University, Linköping, Sweden

3 Division of Otorhinolaryngology, Department of Clinical and Experimental Medicine,

Linköping University, Linköping, Sweden

Correspondence: Ann-Charlotte Johansson, Division of Otorhinolaryngology, Linköping

University Hospital, SE-581 85 Linköping, Sweden

Phone: +46-13-221525, Fax: +46-13-221529, E-mail: [email protected]

Page 3: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

ABSTRACT

Lysosomal membrane permeabilization (LMP) occurs in response to a large variety of cell

death stimuli causing release of cathepsins from the lysosomal lumen into the cytosol where

they participate in apoptosis signaling. In some settings, apoptosis induction is dependent on

an early release of cathepsins, while under other circumstances LMP occurs late in the cell

death process and contributes to amplification of the death signal. The mechanism underlying

LMP is still incompletely understood; however, a growing body of evidence suggests that

LMP may be governed by several distinct mechanisms that are likely engaged in a death

stimulus- and cell-type-dependent fashion. In this review, factors contributing to

permeabilization of the lysosomal membrane including reactive oxygen species, lysosomal

membrane lipid composition, proteases, p53, and Bcl-2 family proteins, are described.

Potential mechanisms to safeguard lysosomal integrity and confer resistance to lysosome-

dependent cell death are also discussed.

Page 4: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

KEYWORDS

Lysosome, Lysosomal release, Caspases, Calpains, Hsp, LAMP

Page 5: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

ABBREVIATIONS

Ahr, aryl hydrocarbon receptor; ANT, adenine nucleotide translocator; apoE, apolipoprotein

E; BH3, Bcl-2 homology 3; Hsp, heat shock protein; JNK, c-Jun N-terminal kinase; LAMP,

lysosome-associated membrane protein; LAPF, lysosome-associated apoptosis-inducing

protein containing the pleckstrin homology and FYVE domains; LEDGF, lens epithelium-

derived growth factor; LMP, lysosomal membrane permeabilization; PI3K,

phosphatidylinositol-3-kinase; PLA2, phospholipase A2; ROS, reactive oxygen species;

siRNA, small-interfering RNA; SMase, sphingomyelinase; TNF, tumor necrosis factor;

TRAIL, tumor necrosis factor-related apoptosis inducing ligand; UV, ultraviolet

Page 6: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

INTRODUCTION

Over the last decade, the lysosome has emerged as a significant component of the cellular

death machinery. Lysosomes, which were first described by de Duve and colleagues in 1955

[1], are acidic, single-membrane bound organelles that are present in all eukaryotic cells [2].

The primary function of lysosomes is degradation of macromolecules, and for this purpose,

lysosomes are filled with more than 50 acid hydrolases, including phosphatases, nucleases,

glycosidases, proteases, peptidases, sulphatases, and lipases [2]. Of the lysosomal hydrolases,

the cathepsin family of proteases is the best characterized. The cathepsins are subdivided,

according to their active site amino acids, into cysteine (cathepsins B, C, F, H, K, L, O, S, V,

W, and X), serine (cathepsins A and G), and aspartic cathepsins (cathepsins D and E) [3].

Cell death through apoptosis is tightly controlled by changes in protein expression, protein-

protein interactions, and various post-translational modifications, including proteolytic

cleavage and phosphorylation. For some of these events to occur, proteins must re-localize

from one sub-cellular compartment to another to gain access to their substrates or interacting

partners. Thus, compartmentalization is yet another important regulatory mechanism,

preventing accidental triggering of cell death signaling. The most obvious example is the

release of apoptogenic factors from mitochondria, a key event in the execution of cell death,

which is regulated mainly by proteins of the Bcl-2 family. Lysosomal hydrolytic enzymes

were initially thought to inevitably trigger necrotic cell death when released into the cytosol

[4]. However, in 1998, we discovered that the aspartic protease cathepsin D was redistributed

from lysosomes to the cytosol upon oxidative stress-induced apoptotic cell death [5] (Figure

1). Cathepsin D was the first identified lysosomal protein with apoptogenic properties, but the

subcellular location of the protein in dying cells was not addressed in the early publications

[6, 7]. In a series of papers from Brunk and co-workers, it was established that the extent of

lysosomal damage determines the cell fate; a limited lysosomal release results in cell death by

apoptosis, while massive lysosomal break-down leads to necrosis [8-12].

It is now generally accepted that apoptosis is often associated with release of cathepsins into

the cytosol, and that, in addition to cathepsin D, cysteine cathepsins B and L are involved in

apoptosis signaling [13-17]. Moreover, lysosomal destabilization, which will subsequently be

referred to as lysosomal membrane permeabilization (LMP), may also occur during other

types of cell death, including necroptosis and autophagic cell death [18, 19]. LMP can be

triggered by a wide range of apoptotic stimuli, including death receptor activation,

Page 7: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

Figure 1. Apoptosis-associated release of lysosomal cathepsin D into the cytosol

Visualization of cathepsin D in rat cardiomyocytes exposed to the redox-cycling drug

naphthazarin by immunofluorescence microscopy (A and B) or immuno-electron microscopy

using antibodies tagged with ultra-small gold particles and subsequent silver enhancement (C

and D). There is a shift from a punctate lysosomal staining pattern in control cells (A) to a

diffuse cytosolic staining in cells exposed to naphthazarin (B). Likewise, in electron

micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows),

while in cells treated with naphthazarin (D), cathepsin D is spread throughout the cytosol

(arrow heads), bars = 30 µm (A and B) and 1 µm (C and D).

endoplasmic reticulum stress, proteasome inhibition, oxidative stress, DNA damage, osmotic

stress, and growth factor starvation [13-17]. In some experimental settings, lysosomal release

is an early event essential for apoptosis signaling to proceed, while under other circumstances

LMP occurs late in the cell death process and contributes to amplification of the death signal

(Figure 2). Cathepsin relocalization seems to be critical for apoptogenicity, and it has been

demonstrated that microinjection of cathepsins into the cytosol is sufficient to trigger

apoptotic cell death [20-22]. Cathepsin release may result in caspase-dependent or -

Page 8: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

independent cell death with or without involvement of mitochondria [13, 17]. Caspases-2 and

-8, phospholipase A2 (PLA2), sphingosine kinase-1, and the Bcl-2 family proteins Bid, Bcl-2,

Bcl-XL, and Mcl-1 may all be downstream targets of cathepsins. A detailed description of the

mechanisms by which lysosomal cathepsins propagate the apoptotic signal can be found

elsewhere [13-16].

Figure 2. Lysosomal participation in cell death signaling

Lysosomal membrane permeabilization (LMP), i.e., release of cathepsins from lysosomes into

the cytosol, is an important step in cell death signaling induced by a variety of stimuli, such as

death receptor activation, radiation, and cytotoxic drugs. Several cytosolic cathepsin targets

have been described and the signaling downstream LMP involves both caspase-dependent and

-independent pathways. Engagement of the mitochondrial pathway is a common downstream

event of LMP, but cathepsins may also cause cell death without involvement of mitochondria.

It is still unclear whether a subset of lysosomes is particularly susceptible to apoptosis-

associated LMP and thus release all of their contents while other lysosomes release none, or if

all lysosomes within a given cell are equally affected but release only part of their contents.

There are, however, indications that interlysosomal differences regarding vulnerability to

Page 9: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

LMP do exist [23, 24]. For example, large lysosomes appears to be more susceptible to LMP

than small ones [24].

The mechanism underlying LMP is still incompletely understood. Apart from cathepsins,

other hydrolases [25-28], H+ (causing acidification of the cytosol) [29], Ca

2+ [30], and several

of the dyes that accumulate in lysosomes with intact membranes, e.g., acridine orange [8],

Lucifer Yellow [10], and LysoTracker [31], can escape into the cytosol at the onset of

apoptosis. This may suggests a nonspecific release mechanism, such as pore formation or

limited membrane damage, rather than selective transport across the membrane, which would

likely be specific for a single group of molecules. However, it should be noted that the release

of H+, Ca

2+, and lysosomotropic dyes does not necessarily reflect a change in membrane

integrity, but could merely be due to a change in ion pump activity. It has been suggested that

apoptosis-associated lysosomal release is size-selective, as 10- and 40-kDa FITC-dextran

molecules are released from lysosomes during staurosporine-induced T-cell apoptosis, while

70- and 250-kDa FITC-dextran molecules are retained [32]. Nevertheless, that upper limit of

size-selection does not apply in all death models, as release of the 150 kDa lysosomal protein

N-acetyl-β-glucosaminidase has been observed under other experimental conditions [25-27].

Conceivably, LMP may be governed by several distinct mechanisms, each with its own

characteristics, which are likely engaged in a death stimulus- and cell-type-dependent fashion.

The mechanisms that have been suggested to contribute to permeabilization of the lysosomal

membrane are described in the following sections, as are those that are thought to safeguard

lysosomal integrity and confer resistance to lysosome-dependent cell death.

PROMOTERS OF LYSOSOMAL MEMBRANE PERMEABILIZATION

Viral and bacterial proteins

Apoptosis triggered by bacterial and viral infections sometimes involves LMP, and in certain

contexts the participation of cathepsins is required for apoptotic signaling to proceed [33-38].

The human immunodeficiency virus type 1 (HIV-1) protein Nef is one of the non-mammalian

proteins that, when entering mammalian cells, causes permeabilization of the lysosomal

membrane [36]. Overexpression of Nef elicits an apoptotic response in CD4+

T lymphocytes

via disruption of lysosomal integrity, thus, the progressive and massive destruction of CD4+

T

lymphocytes characteristic of HIV-1-related disease may in part be due to Nef-induced LMP.

Parvovirus H1 is another virus that induces cell death characterized by translocation of

cathepsins to the cytosol [34]. A small amount of viral proteins were found within the

Page 10: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

lysosomal fraction, and it was speculated that the capsid protein VP1, which possesses a

domain displaying phospholipase A2 (PLA2) activity, could be involved in induction of LMP.

The diphtheria toxin is one of the most interesting examples of a bacterial protein that can

permeabilize the membranes of the endolysosomal compartment. After entering the cell via

endocytosis, one of the subdomains of the toxin inserts into the endosomal membrane and

forms pores through which the catalytic subunit of the toxin passes into the cytosol [39-42]. In

the same way, components of the tetanus [43], botulinum [44], and anthrax [45] toxins are

believed to permeabilize the endosomal membrane to gain access to the cytosol. As discussed

below, certain mammalian proteins involved in apoptosis signaling share high sequence

homology with these viral proteins, and may well be key regulators of apoptosis-associated

LMP.

Pro-apoptotic Bcl-2 family proteins

The Bcl-2 family consists of both pro- (e.g., Bax, Bak, Bid, Bad, Bim, Noxa, and Puma) and

anti-apoptotic (e.g., Bcl-2, Bcl-XL, and Mcl-1) proteins that are critical regulators of the

mitochondrial pathway to cell death. The multi-domain proteins Bax and Bak are pore-

forming proteins that enable the release of apoptogenic factors such as cytochrome c from

mitochondria, while the so-called BH3-only-domain proteins (e.g., Bid, Bad, Bim, Noxa, and

Puma) serve to promote activation of these multi-domain family members upon an apoptotic

insult. The main function of the anti-apoptotic Bcl-2 family proteins, on the other hand, is to

prevent the release of apoptogenic factors. Interestingly, the hypothesis that Bcl-2 family

proteins can release cytochrome c through pore formation was based on the discovery that

some of these proteins showed high structural similarity to the pore-forming domain of the

diphtheria toxin, which, as mentioned above, is known to form pores in the endosomal

membrane [46, 47]. Several years later, we discovered that Bax could also permeabilize the

lysosomal membrane, and thereby promote cell death by the release of lysosomal cathepsins

to the cytosol [26]. Confocal and immuno-electron microscopy showed that Bax translocated

from the cytosol to the lysosomal membrane upon staurosporine treatment of human

fibroblasts. Furthermore, recombinant Bax was inserted into the membrane of isolated rat

liver lysosomes and caused the release of lysosomal constituents. These data provided the first

evidence that Bax is mechanistically involved in LMP and are now supported by a growing

number of publications demonstrating involvement of multiple pro- and anti-apoptotic Bcl-2

family proteins in the regulation of lysosomal membrane integrity.

Page 11: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

Bax is activated and relocated to lysosomes in hepatocytes upon treatment with free fatty

acids [48, 49], in cholangiocarcinoma cells in response to tumor necrosis factor-related

apoptosis inducing ligand (TRAIL) [50], and in ovarian cancer cells treated with etoposide

[51]. In all three cases, small interfering RNA- (siRNA)-mediated downregulation of Bax

prevented LMP, indicating a mechanistic importance of Bax’s lysosomal location.

Furthermore, siRNA-mediated downregulation of Bim was found to prevent TRAIL-induced

LMP [50]. Bim was activated upon TRAIL treatment and colocalized with Bax in lysosomes,

suggesting that it may trigger Bax-mediated LMP. In another study, Bid, the most well-known

Bax activator, was found to be involved in tumor necrosis factor (TNF)-α-induced lysosomal

destabilization in hepatocytes [52]. Upon TNF-α treatment, Bid was detected in lysosomes,

and cathepsin B was released to the cytosol. However, no such release was seen in Bid-/-

cells.

Even though the involvement of Bax was not considered in this study, it is tempting to

speculate that LMP was caused by Bid-mediated activation of Bax. In a recent report, BH3

domain peptides were found to permeabilize isolated lysosomes in the presence of Bax [50].

Interestingly, Bak, the other main pore-forming Bcl-2 family protein, also localizes to

lysosomes [48], but there are as yet no reports of Bak-induced LMP. Conceivably, triggering

of LMP could be a general function of pro-apoptotic Bcl-2 proteins. Similar to mitochondrial

membrane permeabilization [53], LMP-associated Bax activation seems to rely on different

BH3-only proteins depending on the death stimulus and cell type. In addition to a direct effect

of Bcl-2 proteins at the lysosomal membrane, Bcl-2 family members may regulate LMP at the

mitochondrial level as engagement of mitochondria can lead to increased lysosomal release

via positive feed-back. However, as release of cathepsins has been observed in Bax/Bak

double knockout cells there are clearly additional mechanisms independent of Bax and Bak

that contribute to LMP [54].

p53

Permeabilization of the lysosomal membrane occurs in p53-induced apoptosis [55], during

which p53 itself localizes to lysosomes and triggers LMP in a transcription-independent

manner [56]. It was proposed that p53 phosphorylated at Ser15 (p-p53Ser15

) may be recruited

to the lysosomal membrane by a newly discovered protein called Lysosome-associated

apoptosis-inducing protein containing the pleckstrin homology and FYVE domains (LAPF).

This protein, which associates with lysosomes upon induction of apoptosis, is essential for

cell death induced by TNF-α, ionizing irradiation, and the DNA-damaging drugs 5-

fluorouracil and oxaliplatin [56, 57]. Downregulation of either p53 or LAPF prevented TNF-

Page 12: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

α-induced LMP [56]. Interestingly, silencing of LAPF expression abrogated lysosomal

translocation of p-p53Ser15

, whereas silencing of p53 expression had no effect on lysosomal

translocation of LAPF, indicating that LAPF may trigger LMP by acting as an adaptor protein

for p-p53Ser15

. Furthermore, p-p53Ser15

localizes to lysosomes in cultured cortical neurons after

exposure to the psychoactive ingredient of marijuana, ∆9-tetrahydrocannabiol [58], or β-

amyloid [59], both of which cause neuronal cell death. Also in these models of apoptosis,

LMP was abrogated by siRNA-mediated downregulation or chemical inhibition of p53. It is

worth noting that p53, under certain circumstances, localizes to the mitochondrial membrane

during apoptosis and can trigger mitochondrial membrane permeabilization by direct

interaction with Bcl-2 family members [60, 61]. It is tempting to speculate that a similar

mechanism governs p53-induced LMP. Additionally, p53 promotes Bax-mediated

mitochondrial membrane permeabilization through transcriptional upregulation of the BH3-

only-domain proteins Puma and Noxa [62-64]. In this way, p53 may engage the lysosomal

pathway without translocating to lysosomes. Recent data from our lab supports this notion;

ultraviolet B- (UVB)-induced, p53-dependent LMP is associated with an increase in Puma

and Noxa expression without the appearance of p53 at the lysosomal membrane [65].

Proteases

CASPASES

There are a number of publications suggesting that caspases, under certain circumstances, are

involved in the release of apoptogenic factors from lysosomes [50, 52, 66-69]. Caspase-8, in

particular, plays a significant role in the engagement of the lysosomal death pathway

following death receptor activation [50, 52, 67-69]. Caspase-8 can trigger the release of

cathepsins from purified lysosomes, and the effect was enhanced by the addition of a

cytosolic fraction [67], indicating that caspase-8-induced LMP is at least partly mediated by

activation of one or several cytosolic substrates. Bid, which presumably would trigger Bax-

mediated LMP, has been presented as a possible candidate [52]. In our hands, neither caspase-

8 nor activated Bid alone permeabilized the membrane of isolated lysosomes [26], suggesting

that additional factors, such as Bax or Bak, are indeed required. In another intriguing study,

caspase-8 cleaved and activated caspase-9 in an apoptosome-independent fashion [68]. By

using genetically modified mouse embryonic fibroblasts, caspase-9 was proven to be

indispensable for triggering LMP, but the exact mode of action remains elusive. Interestingly,

Page 13: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

depletion of Bid did not prevent the loss of lysosomal membrane integrity. These data suggest

that there are at least two different mechanisms by which caspase-8 is able to induce LMP.

In a recent study, caspase-2 activation was shown to be required for LMP during endoplasmic

reticulum stress-induced apoptosis [70]. The effect was not dependent on Bax, ceramide,

Ca2+

, or reactive oxygen species (ROS) production, suggesting a more direct role of caspase-

2. Indeed, caspase-2 has previously been shown to induce cathepsin release from purified

lysosomes [67].

CATHEPSINS

It has been proposed that lysosomal proteases, in particular cathepsin B, may contribute to

their own release [31, 48, 71]. In mouse hepatocytes, lysosomal destabilization evoked by

TNF-α and sphingosine is dependent on cathepsin B, as induction of LMP fails in cathepsin

B-deficient cells [31]. Moreover, chemical inhibition of cathepsin B significantly reduces both

free fatty acid- and TRAIL-induced LMP [48, 71]. However, it is not clear whether intra- or

extralysosomal cathepsin B is involved. As cathepsin B is constitutively active inside

lysosomes, it cannot be sufficient for LMP; however, it may be necessary. Data in favor of

intralysosomal cathepsin B as a mediator of LMP was provided by cell-free experiments in

which sphingosine was found to cause lysosomal release from cathepsin B-containing but not

from cathepsin B-deficient lysosomes [31]. Alternatively, cathepsin B-mediated LMP may

constitute an amplifying feedback loop, in which a small amount of released cathepsin B

triggers more extensive LMP from outside the lysosome. The latter is supported by the

finding that upregulation of serine protease inhibitor 2A, a potent inhibitor of cathepsin B

located in the cytosol and nucleus, reduces lysosomal breakdown [72].

CALPAINS

Calpains catalyze lysosomal membrane destabilization during neuronal cell death [73-76].

Cell death induced by ischaemia or hypochloric acid is associated with an increase in

cytosolic [Ca2+

], activation of calpains, and LMP. Activated µ-calpain localizes to the

lysosomal membrane prior to release of cathepsins, indicating involvement in the triggering of

LMP [74, 75, 77]. In further support of this hypothesis, -calpain can permeabilize the

membrane of isolated lysosomes [67], and pharmacological inhibition of calpains abrogates

LMP [73]. Interestingly, calpain activity correlates with the appearance of p-p53Ser15

at the

lysosomal membrane, suggesting that calpains may, under certain circumstances, promote

p53-mediated lysosomal release [59].

Page 14: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

Reactive oxygen species

An increased production of ROS may lead to destabilization of the lysosomal membrane via

massive peroxidation of membrane lipids. Photosensitizers located in lysosomes that generate

singlet oxygen upon radiation have been shown to cause rapid release of cathepsins to the

cytosol [78-80]. Moreover, apoptosis induced by UVA radiation or H2O2, both of which

trigger cell death mainly by generating oxidative stress, involves LMP [12, 65, 81].

Additionally, other apoptosis inducers, e.g., N-(4-hydroxyphenyl)retinamide [82] and

vacuolar ATPase inhibitors [83], cause ROS-dependent LMP. However, in the latter model

systems it is not clear whether leakage over the lysosomal membrane is due to ROS-mediated

membrane damage. Alternatively, alterations in the intracellular redox balance may elicit a

redox-dependent signaling cascade resulting in LMP.

Induction of LMP by lysosomal membrane peroxidation reactions has been extensively

studied in model systems in which the intralysosomal iron content has been manipulated [84-

88]. It is hypothesized that intralysosomal iron, originating from degraded metallo proteins,

reacts with H2O2 and generates highly reactive hydroxyl radicals that initiate peroxidation of

the lysosomal membrane (Figure 3). In an illustrative experiment, macrophages that were

allowed to phagocytose silica particles with high amounts of surface-bound iron suffered from

extensive lysosomal damage, while those that ingested silica particles pretreated with the

pharmacologic iron chelator desferrioxamine displayed only modest lysosomal leakage [89].

In several additional studies it has been established that phagocytosis of iron complexes or

iron-containing proteins increases lysosomal vulnerability, while a reduction in

intralysosomal reactive iron reduces lysosomal leakage and cell death [84-88].

Kinases

Cellular signal transduction involves a wide range of small GTPases and kinases constituting

a complex signaling network that ultimately regulates virtually all cellular events including

cell proliferation, differentiation, and cell death. Ras signaling pathways participate in the

overall regulation of lysosomal function, and phosphatidylinositol-3-kinase (PI3K), one of the

downstream targets of Ras, is indispensable for vesicular transport to lysosomes [90-92]. H-

ras driven transformation of NIH3T3 cells causes increased expression of cysteine cathepsins

and reduced lysosomal stability during TNF-induced cell death [93]. Inhibition of PI3K

sensitizes vascular endothelial cells to cytokine-induced apoptosis by increasing lysosomal

permeability [94]. Furthermore, activation of c-Jun N-terminal kinase (JNK) promotes

Page 15: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

lysosomal breakdown in response to TNF-α, TRAIL, and UVB radiation, possibly via

activation of the BH3-only-domain protein Bim [50, 95, 96]. Additionally, in neurons

exposed to β-amyloid, phosphorylation of p53 at Ser15 is JNK1 dependent [97]. In contrast,

inhibitors of JNK fail to affect lysosomal permeability in H-ras transformed NIH3T3 cells.

Instead, activation of the Raf1-extracellular signal-regulated kinase (-ERK) pathway is

responsible for increased engagement of the lysosomal death pathway [98]. Taken together,

these data suggest that LMP is likely regulated by kinase-dependent signaling events,

although the number of studies so far is limited.

Figure 3. Destabilization of the lysosomal membrane by lipid peroxidation

In response to oxidative stress, increased amounts of hydrogen peroxide can diffuse into the

lysosome. In the lysosomal lumen, the acidic milieu and the presence of low-molecular-

weight iron, derived from degraded iron-containing proteins, promotes the reduction of iron

and the generation of hydroxyl radicals. The hydroxyl radicals induce peroxidation of

membrane lipids and thereby cause leakage of lysosomal constituents into the cytosol.

Changes in lysosomal membrane lipid composition

Obviously, the lysosomal membrane composition plays a key role in the maintenance of

lysosomal integrity. Damage to lysosomal membrane components or changes in the

membrane structure and fluidity could result in lysosomal destabilization.

PLA2, phospholipase C, and sphingomyelinase - enzymes that regulate the membrane lipid

composition - all show increased activity in the presence of high cytosolic [Ca2+

], which is

characteristic of apoptosis. Studies using isolated rat lysosomes have shown that these

enzymes [99-102] as well as the phospholipid products arachidonic acid [103],

lysophosphatidylcholine [104], and phosphatidic acid [105], affect the osmotic sensitivity of

lysosomes, making them more susceptible to osmotic stress.

Page 16: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

PLA2 stands out as a particularly interesting candidate because it is involved in lysosomal

membrane destabilization in several different experimental systems [30, 76, 106-109]. For

example, lysosomes are permeabilized in a PLA2-dependent fashion in response to low

concentrations of H2O2 [106]. In this context, activation of PLA2 is dependent on an early

minor release of lysosomal contents (suggesting an alternative mechanism for the initial

release), and PLA2 engages in an amplifying feed-back loop. Notably, incubation of PLA2

with rat liver lysosomes results in leakage of lysosomal constituents.

Apoptosis-associated LMP may also be mediated by an increase in the sphingolipid

sphingosine [10, 31]. Ceramide is produced from sphingomyelin upon activation of

sphingomyelinases (SMase) and is further processed into sphingosine by ceramidase [110]

(Figure 4). It has been proposed that sphingosine accumulates inside lysosomes, where it

permeabilizes the membrane in a detergent-like fashion, resulting in cell death [10]. Under

certain circumstances, sphingosine-induced LMP is dependent on cathepsin B, as sphingosine

fails to permeabilize lysosomes from cathepsin B-/-

hepatocytes [31]. These data also present

the possibility that sphingosine serves as a general trigger of cathepsin B-mediated

intralysosomal events leading to LMP.

In a recent publication, proteins upregulated in lysosomes at the onset of apoptosis-associated

LMP were identified using a proteomic approach [111]. Interestingly, two of these proteins,

prosaposin and protein kinase Cδ, can lead to activation of the lysosomal enzyme acid SMase.

Protein kinase Cδ activates acid SMase after translocating from the cytosol to the lysosomal

membrane [112], and prosaposin is the precursor of four lysosomal sphingolipid activator

proteins (saposins A-D) that promote acid SMase-mediated sphingomyelin degradation [113].

Other factors

β-AMYLOID

Alzheimer’s disease is an age-related progressive neurodegenerative disease in which β-

amyloid peptides are thought to confer neurotoxicity. Accumulation of β-amyloid in

lysosomes of cultured cells results in LMP-dependent cell death [114, 115]. As β-amyloid is

amphipathic and forms micelle-like aggregates [116, 117], the toxicity of β-amyloid could be

mechanistically similar to that of lysosomotropic detergents, which self-assemble into

micelles within lysosomes [118]. Alternatively, β-amyloid, which, like some of the Bcl-2

family proteins, shows structural similarities to pore-forming bacterial toxins, may trigger

Page 17: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

lysosomal release by creating pores. Indeed, oligomeric β-amyloid can incorporate into

artificial lipid bilayers and form pore-like structures [119, 120].

Figure 4. Accumulation of sphingosine leads to lysosomal membrane permeabilization

An apoptotic stimulus, e.g., tumor necrosis factor- , leads to increased activity of

sphingomyelinase, which catalyzes the formation of ceramide from sphingomyelin. By the

action of ceramidase, ceramide is then converted into sphingosine, which accumulates in

lysosomes and acquires detergent-like properties, leading to lysosomal membrane

permeabilization.

APOLIPOPROTEIN E

Apolipoprotein E (apoE) is involved in lipid transport and one of its three major isoforms,

apoE4, is a known risk factor for Alzheimer’s disease. ApoE4-transfected cells are more

susceptible to lysosomal leakage induced by β-amyloid as compared to untransfected cells or

cells transfected with apoE3 [121]. This effect is dependent on a low intralysosomal pH, as

overexpression of apoE4 fails to potentiate leakage from lysosomes neutralized with

bafilomycin or NH4Cl. Furthermore, at acidic pH, apoE4 binds more avidly to phospholipid

bilayer vesicles and potentiates β-amyloid-induced disruption to a greater extent. It was

suggested that, especially at a low pH, apoE4 is converted into a molten globule capable of

binding and altering membrane stability [122]. Thus, such apoE4-mediated effects on

membrane stability are likely restricted to the lysosomal membrane, and may, under certain

circumstances, play a significant role in the determination of cell fate.

Page 18: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

ARYL HYDROCARBON RECEPTOR

In a murine hepatoma cell line in which TNF-α-induced cell death was found to be

independent of caspase-8, LMP was dependent on the aryl hydrocarbon receptor (Ahr) [123].

This receptor is a ligand-activated transcription factor whose target genes are involved in

many cellular functions, including apoptosis. In addition, it has been suggested that the Ahr

may have ligand-independent activities. Ahr deficiency prevents disruption of lysosomes

induced not only by TNF-α treatment, but also following photodynamic therapy [124].

ANT

The adenine nucleotide translocator (ANT) agonist atractyloside, which triggers opening of

the mitochondrial permeability transition pore and release of mitochondrial cytochrome c,

was shown to also induce release of cathepsin B from isolated lysosomes [125]. This was

accidentally discovered by researchers aiming to identify factors released from a

mitochondrial fraction, apparently contaminated with lysosomes. This finding suggests that

ANT-like proteins may be present in the lysosomal membrane and regulate its permeability;

however, in our hands, only a very high concentration of atractyloside permeabilized the

membrane of isolated rat liver lysosomes [26].

GRANULYSIN

The killer effector molecule granulysin, which, together with perforin and granzymes, is

located in the cytolytic granules of human cytotoxic T-lymphocytes and natural killer cells

was recently found to cause necroptosis (programmed necrosis) via LMP [19]. Granulysin,

which interacts with lipids to cause disruption of membranes, has a broad-spectrum

antimicrobial function and shows potent cytotoxic activity against tumor cells. Granulysin

enters lysosomes of tumor cells and triggers release of cathepsin B, which in turn engages the

mitochondrial pathway via cleavage of Bid. Thus, it appears that our immune system is armed

with a weapon to target lysosomes for the destruction of unwanted cells.

SAFEGUARDS OF LYSOSOMAL MEMBRANE INTEGRITY

It has been suggested that the stability of the lysosomal membrane could be modulated in

different ways, and that this could contribute to the determination of cell fate. An example

from normal physiological conditions is the selection of B-lymphocytes in the germinal

centers. B-lymphocytes with high affinity B-cell receptors are spared from apoptotic cell

death by interaction with follicular dendritic cells, which confer resistance to LMP [69]. The

underlying mechanism was not unraveled in this study, but molecules involved in the

Page 19: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

stabilization of the lysosomal membrane have been identified by others (see below).

Furthermore, cancer cells often show various changes in lysosomal function as well as altered

susceptibility to lysosome-mediated cell death. For example, activation of the Ras or Src

oncogenes is associated with changes in the lysosomal distribution, density, size,

ultrastructure, and content, all of which may influence membrane stability [24, 90, 91, 98].

Heat shock proteins

Heat shock proteins (Hsps) are molecular chaperones essential for cells’ ability to cope with

environmental stress [126]. Interestingly, Hsp70 is found at the lysosomes of many tumor

cells and stressed cells, and has been shown to prevent LMP induced by TNF-α, etoposide,

H2O2, or UVB irradiation [27, 127, 128]. Notably, Hsp70 is frequently overexpressed in

tumors, and downregulation of Hsp70 in breast, pancreatic, or colon cancer cells leads to

LMP and cathepsin-mediated cell death without any external death stimuli [27, 129],

suggesting that the tumorigenic potential of Hsp70 could, in part, be due to stabilization of

lysosomes. Hsp70 may prevent lysosomal membrane permeabilization by scavenging of

lysosomal free iron and blocking of oxidative events [128, 130]. Moreover, it is tempting to

speculate that Hsp70, which has been shown to interact with Bax and prevent its translocation

to mitochondria [131, 132], could interfere with Bax-induced LMP.

In addition, Hsp70-2 can prevent permeabilization of the lysosomal membrane [133]. In this

case, the protective effect depends on Hsp70-2-mediated upregulation of lens epithelium-

derived growth factor (LEDGF). Knockdown of LEDGF in cancer cells induces

destabilization of lysosomal membranes and cathepsin-mediated cell death, while ectopic

expression stabilizes lysosomes and prevents cancer cell death. However, as LEDGF has not

been detected in lysosomes, the protective mechanism remains obscure.

Bcl-2 proteins

Anti-apoptotic members of the Bcl-2 family are well-known regulators of mitochondrial

membrane permeabilization, which, during recent years, have gained attention as possible

modulators of lysosomal membrane stability [49, 50, 66, 134, 135]. Our finding that LMP

may be induced by Bax is supported by the fact that overexpression of Bcl-2 prevents

lysosomal destabilization [66, 134, 135]. In two of these studies, it was suggested that Bcl-2

stabilizes lysosomes by preventing activation of PLA2 [134, 135]. However, it is tempting to

speculate that the protective effect of Bcl-2 is, at least in part, due to neutralization of pro-

apoptotic members of the Bcl-2 family. The theory that anti-apoptotic Bcl-2 proteins are

Page 20: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

safeguards of lysosomal membrane integrity is supported by data from Gregory Gores and co-

workers [49, 50]. In these studies, Mcl-1 and Bcl-XL prevented LMP induced by TRAIL and

free fatty acids, respectively. In both cases, Bax was activated and translocated from the

cytosol to lysosomes upon apoptosis induction. Furthermore, overexpression of Mcl-1 and

Bcl-XL, as well as siRNA-mediated downregulation of Bax, stabilized the lysosomal

membrane.

Antioxidants

As described previously, ROS may compromise the integrity of lysosomes via peroxidation of

membrane lipids. A variety of protective mechanisms have evolved to scavenge ROS and

protect cells from their adverse effects; these are collectively known as the cell’s antioxidant

defense. The defense mechanisms include low-molecular-weight antioxidants, for example

vitamins C and E, coenzyme Q10, and glutathione, as well as antioxidant enzymes, such as

glutathione peroxidase, catalase, and superoxide dismutase [136]. Vitamin E, a lipid soluble

antioxidant that protects from lipid peroxidation [137], can prevent lysosomal release [5, 138].

Moreover, LMP induced by N-(4-hydroxyphenyl) retinamide or inhibitors of the vacuolar

ATPase can be abrogated by increasing the amount of intracellular cysteine, which alters the

redox balance [82, 83].

Furthermore, iron-binding proteins may serve to mitigate oxidant-induced LMP, as

intralysosomal free iron can generate highly reactive hydroxyl radicals, leading to

peroxidation of membrane lipids. Indeed, a number of studies have demonstrated that iron-

binding proteins such as ferritin and metallothionein can reduce lysosomal leakage and cell

death [84, 85, 139]. As mentioned above, also Hsp70 can bind intralysosomal redox-active

iron and may, in that way, protect the lysosomal membrane from free-radical attack and

permeabilization [128, 130].

Cholesterol and sphingomyelin

Cholesterol may play an important role in the maintenance of lysosomal membrane stability.

It has long been known that addition of cholesterol to lysosomes reduces their permeability

[140]. Accordingly, a reduced lysosomal membrane cholesterol level is associated with

increased permeability to protons and potassium ions; this, in turn, causes an osmotic

imbalance and destabilization of the lysosomal membrane [141-143]. Sphingomyelin affects

the membrane fluidity by acting as a scaffold for incorporation of cholesterol [144, 145].

Exogenous sphingomyelin, as well as the sphingomyelin derivative 3-O-

Page 21: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

methylsphingomyelin, have been shown to accumulate in the lysosomal membrane and

protect from lysosomal membrane disruption in response to TNF-α and lysosomal

photosensitizers [78]. Thus, apoptosis-associated conversion of sphingomyelin into

sphingosine (Figure 4), a promoter of LMP, appears to have a dual negative effect on

lysosomal membrane integrity.

Lysosome-associated membrane protein-1 and -2

The lysosomal membrane contains highly glycosylated proteins whose complex intralumenal

carbohydrate side chains form a coat on the inner surface of the membrane. It has been

suggested that these glycoproteins serve as a barrier against the hydrolytic activity of the

lysosomal enzymes and thereby prevent accidental release of lysosomal constituents into the

cytosol. Lysosome-associated membrane protein-1 (LAMP)-1 and -2, which constitute ~50%

of the lysosomal membrane proteins [146, 147], can modulate the sensitivity of the lysosomal

membrane to an apoptotic insult [98]. Oncogene-induced reduction in LAMP-1 and -2 can be

caused by cathepsin B overexpression and is associated with increased susceptibility to LMP

in response to photo-oxidation and the anti-cancer drug siramesine. The authors of that

finding speculate that upregulation of lamp-1 and -2 mRNA, which has been observed in

various human cancers [148, 149], might be an attempt to compensate for the deleterious

effect of the reduced half-life of LAMP-1 and -2 in cancer cells overexpressing lysosomal

cysteine cathepsins.

Page 22: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

CONCLUSION

A growing body of evidence suggests that apoptosis is generally associated with LMP,

occurring either as a triggering early event or an amplifying late event. With the wider

acceptance of lysosomes as part of the signaling cascade leading to apoptotic cell death, the

molecular mechanisms governing LMP have received increased attention. As outlined above,

there are many proteinaceous and non-proteinaceous factors, including Bcl-2 family proteins,

ROS, caspases, cathepsins, cholesterol, and Hsps, that influence the stability of the lysosomal

membrane, and their individual importance appears to depend on the cell type and death

stimulus (Figure 5). However, the main mechanism responsible for apoptosis-associated

LMP, if any, remains to be identified. A deeper understanding of the regulation of LMP and

other apoptosis signaling events could enable the development of therapies for diseases

associated with excessive or insufficient apoptosis such as neurodegenerative disorders and

cancer, respectively.

Page 23: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

Figure 5. Factors regulating lysosomal membrane permeabilization

This schematic presents a number of factors that may be responsible for lysosomal membrane

permeabilization (LMP). The relative importance of each mechanism likely depends on the

cell type and death stimulus. Mechanisms that are believed to safeguard lysosomal integrity

and protect from lysosome-mediated cell death are shown in red. “Changes in membrane lipid

composition” includes membrane destabilizing factors such as phospholipase A2 and

sphingosine, as well as LPM protective substances e.g., cholesterol and sphingomyelin.

Abbreviations: JNK, c-Jun N-terminal kinase; LAPF, lysosome-associated apoptosis-inducing

protein containing the pleckstrin homology and FYVE domains; Hsp, heat shock protein;

ROS, reactive oxygen species; ANT, adenine nucleotide translocator; Ahr, aryl hydrocarbon

receptor; LEDGF, lens epithelium-derived growth factor; LAMP, lysosome-associated

membrane protein.

Page 24: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

ACKNOWLEDGEMENTS

We thank Lotta Hellström and Fredrik Jerhammar for critical reading of the manuscript.

Page 25: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

REFERENCES

1. de Duve C, Pressman BC, Gianetto R, Wattiaux R, Applemans F (1955) Tissue

fractionation studies. 6. Intracellular distribution patterns of enzymes in rat-liver

tissue. Biochem J 60:604-617

2. de Duve C (1983) Lysosomes revisited. Eur J Biochem 137:391-397

3. Turk B, Stoka V (2007) Protease signalling in cell death: caspases versus cysteine

cathepsins. FEBS Lett 581:2761-2767

4. de Duve C. Lysosomes, a new group of cytoplasmic particles. In: Hayashi T, ed.

Subcellular particles. New York: Ronald Press 1959: 128-159.

5. Roberg K, Öllinger K (1998) Oxidative stress causes relocation of the lysosomal

enzyme cathepsin D with ensuing apoptosis in neonatal rat cardiomyocytes. Am J

Pathol 152:1151-1156

6. Deiss LP, Galinka H, Berissi H, Cohen O, Kimchi A (1996) Cathepsin D protease

mediates programmed cell death induced by interferon- Fas/APO-1 and TNF- .

EMBO J 15:3861-3870

7. Wu GS, Saftig P, Peters C, El-Deiry WS (1998) Potential role for cathepsin D in p53-

dependent tumor suppression and chemosensitivity. Oncogene 16:2177-2183

8. Brunk UT, Dalen H, Roberg K, Hellquist HB (1997) Photo-oxidative disruption of

lysosomal membranes causes apoptosis of cultured human fibroblasts. Free Radic Biol

Med 23:616-626

9. Brunk UT, Svensson I (1999) Oxidative stress, growth factor starvation and Fas

activation may all cause apoptosis through lysosomal leak. Redox Rep 4:3-11

10. Kågedal K, Zhao M, Svensson I, Brunk UT (2001) Sphingosine-induced apoptosis is

dependent on lysosomal proteases. Biochem J 359:335-343

11. Li W, Yuan XM, Nordgren G et al (2000) Induction of cell death by the

lysosomotropic detergent MSDH. FEBS Lett 470:35-39

12. Antunes F, Cadenas E, Brunk UT (2001) Apoptosis induced by exposure to a low

steady-state concentration of H2O2 is a consequence of lysosomal rupture. Biochem J

356:549-555

13. Boya P, Kroemer G (2008) Lysosomal membrane permeabilization in cell death.

Oncogene 27:6434-6451

14. Chwieralski CE, Welte T, Bühling F (2006) Cathepsin-regulated apoptosis. Apoptosis

11:143-149

15. Guicciardi ME, Leist M, Gores GJ (2004) Lysosomes in cell death. Oncogene

23:2881-2890

16. Stoka V, Turk V, Turk B (2007) Lysosomal cysteine cathepsins: signaling pathways in

apoptosis. Biol Chem 388:555-560

17. Kirkegaard T, Jäättelä M (2009) Lysosomal involvement in cell death and cancer.

Biochim Biophys Acta 1793:746-754

18. Kroemer G, Jaattela M (2005) Lysosomes and autophagy in cell death control. Nat

Rev Cancer 5:886-897

19. Zhang H, Zhong C, Shi L, Guo Y, Fan Z (2009) Granulysin induces cathepsin B

release from lysosomes of target tumor cells to attack mitochondria through

processing of bid leading to Necroptosis. J Immunol 182:6993-7000

20. Roberg K, Kågedal K, Öllinger K (2002) Microinjection of cathepsin D induces

caspase-dependent apoptosis in fibroblasts. Am J Pathol 161:89-96

Page 26: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

21. Schestkowa O, Geisel D, Jacob R, Hasilik A (2007) The catalytically inactive

precursor of cathepsin D induces apoptosis in human fibroblasts and HeLa cells. J Cell

Biochem 101:1558-1566

22. Bivik CA, Larsson PK, Kågedal KM, Rosdahl IK, Öllinger KM (2006) UVA/B-

induced apoptosis in human melanocytes involves translocation of cathepsins and Bcl-

2 family members. J Invest Dermatol 126:1119-1127

23. Nilsson E, Ghassemifar R, Brunk UT (1997) Lysosomal heterogeneity between and

within cells with respect to resistance against oxidative stress. Histochem J 29:857-

865

24. Ono K, Kim SO, Han J (2003) Susceptibility of lysosomes to rupture is a determinant

for plasma membrane disruption in tumor necrosis factor alpha-induced cell death.

Mol Cell Biol 23:665-676

25. Blomgran R, Zheng L, Stendahl O (2007) Cathepsin-cleaved Bid promotes apoptosis

in human neutrophils via oxidative stress-induced lysosomal membrane

permeabilization. J Leukoc Biol 81:1213-1223

26. Kågedal K, Johansson AC, Johansson U et al (2005) Lysosomal membrane

permeabilization during apoptosis-involvement of Bax? Int J Exp Pathol 86:309-321

27. Nylandsted J, Gyrd-Hansen M, Danielewicz A et al (2004) Heat shock protein 70

promotes cell survival by inhibiting lysosomal membrane permeabilization. J Exp

Med 200:425-435

28. Miao Q, Sun Y, Wei T et al (2008) Chymotrypsin B cached in rat liver lysosomes and

involved in apoptotic regulation through a mitochondrial pathway. J Biol Chem

283:8218-8228

29. Nilsson C, Johansson U, Johansson AC, Kågedal K, Öllinger K (2006) Cytosolic

acidification and lysosomal alkalinization during TNF-alpha induced apoptosis in

U937 cells. Apoptosis 11:1149-1159

30. Mirnikjoo B, Balasubramanian K, Schroit AJ (2009) Mobilization of lysosomal

calcium regulates the externalization of phosphatidylserine during apoptosis. J Biol

Chem 284:6918-6923

31. Werneburg NW, Guicciardi ME, Bronk SF, Gores GJ (2002) Tumor necrosis factor-

alpha-associated lysosomal permeabilization is cathepsin B dependent. Am J Physiol-

Gastroint Liver Physiol 283:G947-956

32. Bidère N, Lorenzo HK, Carmona S et al (2003) Cathepsin D triggers Bax activation,

resulting in selective apoptosis-inducing factor (AIF) relocation in T lymphocytes

entering the early commitment phase to apoptosis. J Biol Chem 278:31401-31411

33. Birk AV, Dubovi EJ, Cohen-Gould L, Donis R, Szeto HH (2008) Cytoplasmic

vacuolization responses to cytopathic bovine viral diarrhoea virus. Virus Res 132:76-

85

34. Di Piazza M, Mader C, Geletneky K et al (2007) Cytosolic activation of cathepsins

mediates parvovirus H-1-induced killing of cisplatin and TRAIL-resistant glioma

cells. J Virol 81:4186-4198

35. Kaznelson DW, Bruun S, Monrad A et al (2004) Simultaneous human papilloma virus

type 16 E7 and cdk inhibitor p21 expression induces apoptosis and cathepsin B

activation. Virology 320:301-312

36. Laforge M, Petit F, Estaquier J, Senik A (2007) Commitment to apoptosis in CD4+ T

lymphocytes productively infected with human immunodeficiency virus type 1 is

initiated by lysosomal membrane permeabilization, itself induced by the isolated

expression of the viral protein Nef. J Virol 81:11426-11440

Page 27: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

37. O'Sullivan MP, O'Leary S, Kelly DM, Keane J (2007) A caspase-independent pathway

mediates macrophage cell death in response to Mycobacterium tuberculosis infection.

Infect Immun 75:1984-1993

38. Prince LR, Bianchi SM, Vaughan KM et al (2008) Subversion of a lysosomal pathway

regulating neutrophil apoptosis by a major bacterial toxin, pyocyanin. J Immunol

180:3502-3511

39. Sandvig K (2003) Transport of toxins across intracellular membranes. In: Burns DL,

Barbier JT, Iglewski BH (eds) Bacterial protein toxins. ASM, Washington, DC, pp

157-172

40. Sandvig K, Olsnes S (1980) Diphtheria toxin entry into cells is facilitated by low pH. J

Cell Biol 87:828-832

41. Donovan JJ, Simon MI, Draper RK, Montal M (1981) Diphtheria toxin forms

transmembrane channels in planar lipid bilayers. Proc Natl Acad Sci U S A 78:172-

176

42. Kagan BL, Finkelstein A, Colombini M (1981) Diphtheria toxin fragment forms large

pores in phospholipid bilayer membranes. Proc Natl Acad Sci U S A 78:4950-4954

43. Boquet P, Duflot E (1982) Tetanus toxin fragment forms channels in lipid vesicles at

low pH. Proc Natl Acad Sci U S A 79:7614-7618

44. Donovan JJ, Middlebrook JL (1986) Ion-conducting channels produced by botulinum

toxin in planar lipid membranes. Biochemistry 25:2872-2876

45. Blaustein RO, Koehler TM, Collier RJ, Finkelstein A (1989) Anthrax toxin: channel-

forming activity of protective antigen in planar phospholipid bilayers. Proc Natl Acad

Sci U S A 86:2209-2213

46. Muchmore SW, Sattler M, Liang H et al (1996) X-ray and NMR structure of human

Bcl-xL, an inhibitor of programmed cell death. Nature 381:335-341

47. Suzuki M, Youle RJ, Tjandra N (2000) Structure of Bax: coregulation of dimer

formation and intracellular localization. Cell 103:645-654

48. Feldstein AE, Werneburg NW, Canbay A et al (2004) Free fatty acids promote hepatic

lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway.

Hepatology 40:185-194

49. Feldstein AE, Werneburg NW, Li Z, Bronk SF, Gores GJ (2006) Bax inhibition

protects against free fatty acid-induced lysosomal permeabilization. Am J Physiol-

Gastroint Liver Physiol 290:G1339-1346

50. Werneburg NW, Guicciardi ME, Bronk SF, Kaufmann SH, Gores GJ (2007) Tumor

necrosis factor-related apoptosis-inducing ligand activates a lysosomal pathway of

apoptosis that is regulated by Bcl-2 proteins. J Biol Chem 282:28960-28970

51. Castino R, Peracchio C, Salini A et al (2009) Chemotherapy Drug Response in

Ovarian Cancer Cells Strictly Depends on a Cathepsin D - Bax Activation Loop. J Cell

Mol Med 13:1096-1109

52. Werneburg NW, Guicciardi ME, Yin XM, Gores GJ (2004) TNF-alpha-mediated

lysosomal permeabilization is FAN and caspase 8/Bid dependent. Am J Physiol-

Gastroint Liver Physiol 287:G436-443

53. Youle RJ, Strasser A (2008) The BCL-2 protein family: opposing activities that

mediate cell death. Nat Rev Mol Cell Biol 9:47-59

54. Boya P, Andreau K, Poncet D et al (2003) Lysosomal membrane permeabilization

induces cell death in a mitochondrion-dependent fashion. J Exp Med 197:1323-1334

55. Yuan XM, Li W, Dalen H et al (2002) Lysosomal destabilization in p53-induced

apoptosis. Proc Natl Acad Sci U S A 99:6286-6291

Page 28: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

56. Li N, Zheng Y, Chen W et al (2007) Adaptor protein LAPF recruits phosphorylated

p53 to lysosomes and triggers lysosomal destabilization in apoptosis. Cancer Res

67:11176-11185

57. Chen W, Li N, Chen T et al (2005) The lysosome-associated apoptosis-inducing

protein containing the pleckstrin homology (PH) and FYVE domains (LAPF),

representative of a novel family of PH and FYVE domain-containing proteins, induces

caspase-independent apoptosis via the lysosomal-mitochondrial pathway. J Biol Chem

280:40985-40995

58. Gowran A, Campbell VA (2008) A role for p53 in the regulation of lysosomal

permeability by delta 9-tetrahydrocannabinol in rat cortical neurones: implications for

neurodegeneration. J Neurochem 105:1513-1524

59. Fogarty MP, McCormack RM, Noonan J, Murphy D, Gowran A, Campbell VA

(2008) A role for p53 in the beta-amyloid-mediated regulation of the lysosomal

system. Neurobiol Aging Doi:10.1016/j.neurobiolaging.2008.09.018

60. Chipuk JE, Kuwana T, Bouchier-Hayes L et al (2004) Direct activation of Bax by p53

mediates mitochondrial membrane permeabilization and apoptosis. Science 303:1010-

1014

61. Mihara M, Erster S, Zaika A et al (2003) p53 has a direct apoptogenic role at the

mitochondria. Mol Cell 11:577-590

62. Oda E, Ohki R, Murasawa H et al (2000) Noxa, a BH3-only member of the Bcl-2

family and candidate mediator of p53-induced apoptosis. Science 288:1053-1058

63. Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B (2001) PUMA induces the

rapid apoptosis of colorectal cancer cells. Mol Cell 7:673-682

64. Nakano K, Vousden KH (2001) PUMA, a novel proapoptotic gene, is induced by p53.

Mol Cell 7:683-694

65. Wäster PK, Öllinger KM (2009) Redox-dependent translocation of p53 to

mitochondria or nucleus in human melanocytes after UVA- and UVB-induced

apoptosis. J Invest Dermatol 129:1769-1781

66. Paris C, Bertoglio J, Bréard J (2007) Lysosomal and mitochondrial pathways in

miltefosine-induced apoptosis in U937 cells. Apoptosis 12:1257-1267

67. Guicciardi ME, Deussing J, Miyoshi H et al (2000) Cathepsin B contributes to TNF-

-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c.

J Clin Invest 106:1127-1137

68. Gyrd-Hansen M, Farkas T, Fehrenbacher N et al (2006) Apoptosome-independent

activation of the lysosomal cell death pathway by caspase-9. Mol Cell Biol 26:7880-

7891

69. van Nierop K, Muller FJ, Stap J, Van Noorden CJ, van Eijk M, de Groot C (2006)

Lysosomal destabilization contributes to apoptosis of germinal center B-lymphocytes.

J Histochem Cytochem 54:1425-1435

70. Huang WC, Lin YS, Chen CL, Wang CY, Chiu WH, Lin CF (2009) Glycogen

synthase kinase-3beta mediates endoplasmic reticulum stress-induced lysosomal

apoptosis in leukemia. J Pharmacol Exp Ther 329:524-531

71. Guicciardi ME, Bronk SF, Werneburg NW, Gores GJ (2007) cFLIPL prevents TRAIL-

induced apoptosis of hepatocellular carcinoma cells by inhibiting the lysosomal

pathway of apoptosis. Am J Physiol-Gastroint Liver Physiol 292:G1337-1346

72. Liu N, Raja SM, Zazzeroni F et al (2003) NF-kappaB protects from the lysosomal

pathway of cell death. EMBO J 22:5313-5322

73. Yap YW, Whiteman M, Bay BH et al (2006) Hypochlorous acid induces apoptosis of

cultured cortical neurons through activation of calpains and rupture of lysosomes. J

Neurochem 98:1597-1609

Page 29: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

74. Yamashima T, Saido TC, Takita M et al (1996) Transient brain ischaemia provokes

Ca2+, PIP2 and calpain responses prior to delayed neuronal death in monkeys. Eur J

Neurosci 8:1932-1944

75. Yamashima T, Tonchev AB, Tsukada T et al (2003) Sustained calpain activation

associated with lysosomal rupture executes necrosis of the postischemic CA1 neurons

in primates. Hippocampus 13:791-800

76. Windelborn JA, Lipton P (2008) Lysosomal release of cathepsins causes ischemic

damage in the rat hippocampal slice and depends on NMDA-mediated calcium influx,

arachidonic acid metabolism, and free radical production. J Neurochem 106:56-69

77. Yamashima T, Kohda Y, Tsuchiya K et al (1998) Inhibition of ischaemic hippocampal

neuronal death in primates with cathepsin B inhibitor CA-074: a novel strategy for

neuroprotection based on 'calpain-cathepsin hypothesis'. Eur J Neurosci 10:1723-1733

78. Caruso JA, Mathieu PA, Reiners JJ, Jr. (2005) Sphingomyelins suppress the targeted

disruption of lysosomes/endosomes by the photosensitizer NPe6 during photodynamic

therapy. Biochem J 392:325-334

79. Kessel D, Luo Y, Mathieu P, Reiners JJ, Jr. (2000) Determinants of the apoptotic

response to lysosomal photodamage. Photochem Photobiol 71:196-200

80. Reiners JJ, Jr., Caruso JA, Mathieu P, Chelladurai B, Yin XM, Kessel D (2002)

Release of cytochrome c and activation of pro-caspase-9 following lysosomal

photodamage involves Bid cleavage. Cell Death Differ 9:934-944

81. Öllinger K, Brunk UT (1995) Cellular injury induced by oxidative stress is mediated

through lysosomal damage. Free Radic Biol Med 19:565-574

82. Vene R, Arena G, Poggi A et al (2007) Novel cell death pathways induced by N-(4-

hydroxyphenyl)retinamide: therapeutic implications. Mol Cancer Ther 6:286-298

83. De Milito A, Iessi E, Logozzi M et al (2007) Proton pump inhibitors induce apoptosis

of human B-cell tumors through a caspase-independent mechanism involving reactive

oxygen species. Cancer Res 67:5408-5417

84. Garner B, Li W, Roberg K, Brunk UT (1997) On the cytoprotective role of ferritin in

macrophages and its ability to enhance lysosomal stability. Free Radic Res 27:487-500

85. Persson HL, Nilsson KJ, Brunk UT (2001) Novel cellular defenses against iron and

oxidation: ferritin and autophagocytosis preserve lysosomal stability in airway

epithelium. Redox Rep 6:57-63

86. Persson HL, Yu Z, Tirosh O, Eaton JW, Brunk UT (2003) Prevention of oxidant-

induced cell death by lysosomotropic iron chelators. Free Radic Biol Med 34:1295-

1305

87. Persson HL, Kurz T, Eaton JW, Brunk UT (2005) Radiation-induced cell death:

importance of lysosomal destabilization. Biochem J 389:877-884

88. Yu Z, Persson HL, Eaton JW, Brunk UT (2003) Intralysosomal iron: a major

determinant of oxidant-induced cell death. Free Radic Biol Med 34:1243-1252

89. Persson HL (2005) Iron-dependent lysosomal destabilization initiates silica-induced

apoptosis in murine macrophages. Toxicol Lett 159:124-133

90. Brown WJ, DeWald DB, Emr SD, Plutner H, Balch WE (1995) Role for

phosphatidylinositol 3-kinase in the sorting and transport of newly synthesized

lysosomal enzymes in mammalian cells. J Cell Biol 130:781-796

91. Mousavi SA, Brech A, Berg T, Kjeken R (2003) Phosphoinositide 3-kinase regulates

maturation of lysosomes in rat hepatocytes. Biochem J 372:861-869

92. Fehrenbacher N, Philips M (2009) Intracellular signaling: peripatetic Ras. Curr Biol

19:R454-457

Page 30: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

93. Fehrenbacher N, Gyrd-Hansen M, Poulsen B et al (2004) Sensitization to the

lysosomal cell death pathway upon immortalization and transformation. Cancer Res

64:5301-5310

94. Madge LA, Li J-H, Choi J, Pober JS (2003) Inhibition of phosphatidylinositol 3-

kinase sensitizes vascular endothelial cells to cytokine-initiated cathepsin-dependent

apoptosis. J Biol Chem 278:21295-21206

95. Dietrich N, Thastrup J, Holmberg C et al (2004) JNK2 mediates TNF-induced cell

death in mouse embryonic fibroblasts via regulation of both caspase and cathepsin

protease pathways. Cell Death Differ 11:301-313

96. Bivik C, Öllinger K (2008) JNK mediates UVB-induced apoptosis upstream

lysosomal membrane permeabilization and Bcl-2 family proteins. Apoptosis 13:1111-

1120

97. Fogarty MP, Downer EJ, Campbell V (2003) A role for c-Jun N-terminal kinase 1

(JNK1), but not JNK2, in the beta-amyloid-mediated stabilization of protein p53 and

induction of the apoptotic cascade in cultured cortical neurons. Biochem J 371:789-

798

98. Fehrenbacher N, Bastholm L, Kirkegaard-Sorensen T et al (2008) Sensitization to the

lysosomal cell death pathway by oncogene-induced down-regulation of lysosome-

associated membrane proteins 1 and 2. Cancer Res 68:6623-6633

99. Zhao HF, Wang X, Zhang GJ (2005) Lysosome destabilization by cytosolic extracts,

putative involvement of Ca(2+)/phospholipase C. FEBS Lett 579:1551-1556

100. Wang X, Zhao HF, Zhang GJ (2006) Mechanism of cytosol phospholipase C and

sphingomyelinase-induced lysosome destabilization. Biochimie 88:913-922

101. Wang JW, Sun L, Hu JS, Li YB, Zhang GJ (2006) Effects of phospholipase A2 on the

lysosomal ion permeability and osmotic sensitivity. Chem Phys Lipids 144:117-126

102. Wang X, Wang LL, Zhang GJ (2006) Guanosine 5'-[gamma-thio]triphosphate-

mediated activation of cytosol phospholipase C caused lysosomal destabilization. J

Membr Biol 211:55-63

103. Zhang G, Yi YP, Zhang GJ (2006) Effects of arachidonic acid on the lysosomal ion

permeability and osmotic stability. J Bioenerg Biomembr 38:75-82

104. Hu JS, Li YB, Wang JW, Sun L, Zhang GJ (2007) Mechanism of

lysophosphatidylcholine-induced lysosome destabilization. J Membr Biol 215:27-35

105. Yi YP, Wang X, Zhang G, Fu TS, Zhang GJ (2006) Phosphatidic acid osmotically

destabilizes lysosomes through increased permeability to K+ and H+. Gen Physiol

Biophys 25:149-160

106. Zhao M, Brunk UT, Eaton JW (2001) Delayed oxidant-induced cell death involves

activation of phospholipase A2. FEBS Lett 509:399-404

107. Burlando B, Marchi B, Panfoli I, Viarengo A (2002) Essential role of Ca2+ -

dependent phospholipase A2 in estradiol-induced lysosome activation. Am J Physiol

Cell Physiol 283:C1461-1468

108. Marone G, Fimiani B, Torella G, Poto S, Bianco P, Condorelli M (1983) Possible role

of arachidonic acid and of phospholipase A2 in the control of lysosomal enzyme

release from human polymorphonuclear leukocytes. J Clin Lab Immunol 12:111-116

109. Marchi B, Burlando B, Moore MN, Viarengo A (2004) Mercury- and copper-induced

lysosomal membrane destabilisation depends on [Ca2+]i dependent phospholipase A2

activation. Aquat Toxicol 66:197-204

110. Schutze S, Machleidt T, Adam D et al (1999) Inhibition of receptor internalization by

monodansylcadaverine selectively blocks p55 tumor necrosis factor receptor death

domain signaling. J Biol Chem 274:10203-10212

Page 31: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

111. Parent N, Winstall E, Beauchemin M, Paquet C, Poirier GG, Bertrand R (2009)

Proteomic analysis of enriched lysosomes at early phase of camptothecin-induced

apoptosis in human U-937 cells. J Proteomics 72:960-973

112. Zeidan YH, Hannun YA (2007) Activation of acid sphingomyelinase by protein

kinase Cdelta-mediated phosphorylation. J Biol Chem 282:11549-11561

113. Ferlinz K, Linke T, Bartelsen O, Weiler M, Sandhoff K (1999) Stimulation of

lysosomal sphingomyelin degradation by sphingolipid activator proteins. Chem Phys

Lipids 102:35-43

114. Yang AJ, Chandswangbhuvana D, Shu T, Henschen A, Glabe CG (1999) Intracellular

accumulation of insoluble, newly synthesized abetan-42 in amyloid precursor protein-

transfected cells that have been treated with Abeta1-42. J Biol Chem 274:20650-

20656

115. Zheng L, Kågedal K, Dehvari N et al (2009) Oxidative stress induces macroautophagy

of amyloid beta-protein and ensuing apoptosis. Free Radic Biol Med 46:422-429

116. Soreghan B, Kosmoski J, Glabe C (1994) Surfactant properties of Alzheimer's A beta

peptides and the mechanism of amyloid aggregation. J Biol Chem 269:28551-28554

117. Lomakin A, Chung DS, Benedek GB, Kirschner DA, Teplow DB (1996) On the

nucleation and growth of amyloid beta-protein fibrils: detection of nuclei and

quantitation of rate constants. Proc Natl Acad Sci U S A 93:1125-1129

118. Forster S, Scarlett L, Lloyd JB (1987) The effect of lysosomotropic detergents on the

permeability properties of the lysosome membrane. Biochim Biophys Acta 924:452-

457

119. Lin H, Bhatia R, Lal R (2001) Amyloid beta protein forms ion channels: implications

for Alzheimer's disease pathophysiology. FASEB J 15:2433-2444

120. Quist A, Doudevski I, Lin H et al (2005) Amyloid ion channels: a common structural

link for protein-misfolding disease. Proc Natl Acad Sci U S A 102:10427-10432

121. Ji ZS, Miranda RD, Newhouse YM, Weisgraber KH, Huang Y, Mahley RW (2002)

Apolipoprotein E4 potentiates amyloid beta peptide-induced lysosomal leakage and

apoptosis in neuronal cells. J Biol Chem 277:21821-21828

122. Ji ZS, Mullendorff K, Cheng IH, Miranda RD, Huang Y, Mahley RW (2006)

Reactivity of apolipoprotein E4 and amyloid beta peptide: lysosomal stability and

neurodegeneration. J Biol Chem 281:2683-2692

123. Caruso JA, Mathieu PA, Joiakim A, Zhang H, Reiners JJ, Jr. (2006) Aryl hydrocarbon

receptor modulation of tumor necrosis factor-alpha-induced apoptosis and lysosomal

disruption in a hepatoma model that is caspase-8-independent. J Biol Chem

281:10954-10967

124. Caruso JA, Mathieu PA, Joiakim A et al (2004) Differential susceptibilities of murine

hepatoma 1c1c7 and Tao cells to the lysosomal photosensitizer NPe6: influence of

aryl hydrocarbon receptor on lysosomal fragility and protease contents. Mol

Pharmacol 65:1016-1028

125. Vancompernolle K, Van Herreweghe F, Pynaert G et al (1998) Atractyloside-induced

release of cathepsin B, a protease with caspase-processing activity. FEBS Lett

438:150-158

126. Jäättelä M (1999) Escaping cell death: survival proteins in cancer. Exp Cell Res

248:30-43

127. Bivik C, Rosdahl I, Öllinger K (2007) Hsp70 protects against UVB induced apoptosis

by preventing release of cathepsins and cytochrome c in human melanocytes.

Carcinogenesis 28:537-544

Page 32: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

128. Doulias PT, Kotoglou P, Tenopoulou M et al (2007) Involvement of heat shock

protein-70 in the mechanism of hydrogen peroxide-induced DNA damage: the role of

lysosomes and iron. Free Radic Biol Med 42:567-577

129. Dudeja V, Mujumdar N, Phillips P et al (2009) Heat shock protein 70 inhibits

apoptosis in cancer cells through simultaneous and independent mechanisms.

Gastroenterology 136:1772-1782

130. Kurz T, Brunk UT (2009) Autophagy of HSP70 and chelation of lysosomal iron in a

non-redox-active form. Autophagy 5:93-95

131. Gotoh T, Terada K, Oyadomari S, Mori M (2004) hsp70-DnaJ chaperone pair prevents

nitric oxide- and CHOP-induced apoptosis by inhibiting translocation of Bax to

mitochondria. Cell Death Differ 11:390-402

132. Stankiewicz AR, Lachapelle G, Foo CP, Radicioni SM, Mosser DD (2005) Hsp70

inhibits heat-induced apoptosis upstream of mitochondria by preventing Bax

translocation. J Biol Chem 280:38729-38739

133. Daugaard M, Kirkegaard-Sørensen T, Ostenfeld MS et al (2007) Lens epithelium-

derived growth factor is an Hsp70-2 regulated guardian of lysosomal stability in

human cancer. Cancer Res 67:2559-2567

134. Zhao M, Eaton JW, Brunk UT (2000) Protection against oxidant-mediated lysosomal

rupture: a new anti-apoptotic activity of Bcl-2? FEBS Lett 485:104-108

135. Zhao M, Eaton JW, Brunk UT (2001) Bcl-2 phosphorylation is required for inhibition

of oxidative stress-induced lysosomal leak and ensuing apoptosis. FEBS Lett 509:405-

412

136. Halliwell B (1999) Antioxidant defence mechanisms: from the beginning to the end

(of the beginning). Free Radic Res 31:261-272

137. Yoshida Y, Saito Y, Jones LS, Shigeri Y (2007) Chemical reactivities and physical

effects in comparison between tocopherols and tocotrienols: physiological significance

and prospects as antioxidants. J Biosci Bioeng 104:439-445

138. Mukherjee AK, Ghosal SK, Maity CR (1997) Lysosomal membrane stabilization by

alpha-tocopherol against the damaging action of Vipera russelli venom phospholipase

A2. Cell Mol Life Sci 53:152-155

139. Baird SK, Kurz T, Brunk UT (2006) Metallothionein protects against oxidative stress-

induced lysosomal destabilization. Biochem J 394:275-283

140. Fouchier F, Mego JL, Dang J, Simon C (1983) Thyroid lysosomes: the stability of the

lysosomal membrane. Eur J Cell Biol 30:272-278

141. Deng D, Jiang N, Hao SJ, Sun H, Zhang GJ (2009) Loss of membrane cholesterol

influences lysosomal permeability to potassium ions and protons. Biochim Biophys

Acta 1788:470-476

142. Jadot M, Andrianaivo F, Dubois F, Wattiaux R (2001) Effects of methylcyclodextrin

on lysosomes. Eur J Biochem 268:1392-1399

143. Hao SJ, Hou JF, Jiang N, Zhang GJ (2008) Loss of membrane cholesterol affects

lysosomal osmotic stability. Gen Physiol Biophys 27:278-283

144. Ridgway ND (2000) Interactions between metabolism and intracellular distribution of

cholesterol and sphingomyelin. Biochim Biophys Acta 1484:129-141

145. Simons K, Ikonen E (2000) How cells handle cholesterol. Science 290:1721-1726

146. Eskelinen EL (2006) Roles of LAMP-1 and LAMP-2 in lysosome biogenesis and

autophagy. Mol Aspects Med 27:495-502

147. Fukuda M (1991) Lysosomal membrane glycoproteins. Structure, biosynthesis, and

intracellular trafficking. J Biol Chem 266:21327-21330

Page 33: Regulation of apoptosis-associated lysosomal membrane ...315254/FULLTEXT01.pdf · micrographs (C), cathepsin D can be seen in lysosome-like structures in control cells (arrows), while

148. Furuta K, Ikeda M, Nakayama Y et al (2001) Expression of lysosome-associated

membrane proteins in human colorectal neoplasms and inflammatory diseases. Am J

Pathol 159:449-455

149. Ozaki K, Nagata M, Suzuki M et al (1998) Isolation and characterization of a novel

human lung-specific gene homologous to lysosomal membrane glycoproteins 1 and 2:

significantly increased expression in cancers of various tissues. Cancer Res 58:3499-

3503